HemaMax™, a recombinant human interleukin-12, is a potent mitigator of acute radiation injury in mice and non-human primates

Lena A Basile, Dolph Ellefson, Zoya Gluzman-Poltorak, Katiana Junes-Gill, Vernon Mar, Sarita Mendonca, Joseph D Miller, Jamie Tom, Alice Trinh, Timothy K Gallaher, Lena A Basile, Dolph Ellefson, Zoya Gluzman-Poltorak, Katiana Junes-Gill, Vernon Mar, Sarita Mendonca, Joseph D Miller, Jamie Tom, Alice Trinh, Timothy K Gallaher

Abstract

HemaMax, a recombinant human interleukin-12 (IL-12), is under development to address an unmet medical need for effective treatments against acute radiation syndrome due to radiological terrorism or accident when administered at least 24 hours after radiation exposure. This study investigated pharmacokinetics, pharmacodynamics, and efficacy of m-HemaMax (recombinant murine IL-12), and HemaMax to increase survival after total body irradiation (TBI) in mice and rhesus monkeys, respectively, with no supportive care. In mice, m-HemaMax at an optimal 20 ng/mouse dose significantly increased percent survival and survival time when administered 24 hours after TBI between 8-9 Gy (p<0.05 Pearson's chi-square test). This survival benefit was accompanied by increases in plasma interferon-γ (IFN-γ) and erythropoietin levels, recovery of femoral bone hematopoiesis characterized with the presence of IL-12 receptor β2 subunit-expressing myeloid progenitors, megakaryocytes, and osteoblasts. Mitigation of jejunal radiation damage was also examined. At allometrically equivalent doses, HemaMax showed similar pharmacokinetics in rhesus monkeys compared to m-HemaMax in mice, but more robustly increased plasma IFN-γ levels. HemaMax also increased plasma erythropoietin, IL-15, IL-18, and neopterin levels. At non-human primate doses pharmacologically equivalent to murine doses, HemaMax (100 ng/Kg and 250 ng/Kg) administered at 24 hours after TBI (6.7 Gy/LD(50/30)) significantly increased percent survival of HemaMax groups compared to vehicle (p<0.05 Pearson's chi-square test). This survival benefit was accompanied by a significantly higher leukocyte (neutrophils and lymphocytes), thrombocyte, and reticulocyte counts during nadir (days 12-14) and significantly less weight loss at day 12 compared to vehicle. These findings indicate successful interspecies dose conversion and provide proof of concept that HemaMax increases survival in irradiated rhesus monkeys by promoting hematopoiesis and recovery of immune functions and possibly gastrointestinal functions, likely through a network of interactions involving dendritic cells, osteoblasts, and soluble factors such as IL-12, IFN-γ, and cytoprotectant erythropoietin.

Conflict of interest statement

Competing Interests: Lena A. Basile, Dolph Ellefson, Zoya Gluzman-Poltorak, Katiana Junes-Gill, Vernon Mar, Sarita Mendonca, Jamie Tom, Alice Trinh, and Timothy K. Gallaher are employees of Neumedicines Inc. Joseph D. Miller is a consultant to Neumedicines Inc. HemaMax is a Neumedicines' product; there are numerous relevant patents, details of which are available on request. This does not alter the authors' adherence to all the PLoS ONE policies on sharing data and materials.

Figures

Figure 1. m-HemaMax administered at least 24…
Figure 1. m-HemaMax administered at least 24 hours after TBI increased survival time of irradiated mice.
(a) Animals received vehicle or m-HemaMax at an ostensible dose of 100 ng/mouse at 24 hours and 72 hours after a TBI of 8 Gy (LD86/30). (b) Animals received vehicle or a single, ostensible dose of 300 ng/mouse of m-HemaMax at 24 hours, 48 hours, or 72 hours after a TBI of 9 Gy (LD100/30). (c) Animals received vehicle or a single low dose of m-HemaMax (2 ng/mouse or 18 ng/mouse) at 24 hours after a TBI of 7.9 Gy (LD85/30). Vehicle and m-HemaMax were injected subcutaneously. Vehicle was PBS in (a) and (b) and P5.6TT in (c). The delivered m-HemaMax dose was estimated to be 10 ng/mouse in (a) and 30 ng/mouse in (b) because subsequent studies showed that the actual m-HemaMax dose delivered was approximately 10% of the intended dose, most likely due to m-HemaMax sticking to surfaces of vials and syringes.
Figure 2. Efficacy of m-HemaMax in increasing…
Figure 2. Efficacy of m-HemaMax in increasing survival is not dependent on radiation dose in mice.
Animals were subjected to TBI at ascending radiation doses of 8.6 Gy (LD70/30), 8.8 Gy (LD90/30), and 9.0 Gy (LD100/30) and subsequently received m-HemaMax at a dose of 20 ng/mouse 24 hours after irradiation. Mice were monitored for survival up to day 30. Vehicle was P5.6TT.
Figure 3. m-HemaMax administration increased plasma m-HemaMax…
Figure 3. m-HemaMax administration increased plasma m-HemaMax and IFN-γ levels in irradiated and non-irradiated mice.
Animals received m-HemaMax subcutaneously at a dose of (a) 10 ng/mouse, (b) 20 ng/mouse, (c) 40 ng/mouse, or (d) 200 ng/mouse in the absence of irradiation or at 24 hours after an LD90/30 of TBI. The plasma concentrations of m-HemaMax and IFN-γ were determined by ELISA in blood samples withdrawn at the indicated times. The y-axis scale in (d) is 8 times greater than those in (a) and (b) and 5 times greater than that in (c). n = 3 per timepoint in each group.
Figure 4. Optimal m-HemaMax dose of 20…
Figure 4. Optimal m-HemaMax dose of 20 ng/mouse increased plasma EPO concentration in irradiated mice.
Animals received m-HemaMax subcutaneously at a dose of (a) 10 ng/mouse, (b) 20 ng/mouse, (c) 40 ng/mouse, or (d) 200 ng/mouse in the absence of irradiation or at 24 hours after an LD90/30 of TBI. The plasma concentrations of EPO were determined by ELISA in blood samples withdrawn at 12 hours after m-HemaMax administration.
Figure 5. m-HemaMax promotes hematopoietic recovery in…
Figure 5. m-HemaMax promotes hematopoietic recovery in irradiated mice.
Representative sections of femoral bone marrow from non-irradiated, untreated mice that were stained for IL-12Rβ2 (orange color) are shown in (a). Animals were subjected to TBI (8.0 Gy) and subsequently received vehicle (P5.6TT) or m-HemaMax (20 ng/mouse) subcutaneously at the indicated times post irradiation (b–f). An additional group of mice received HemaMax at 24 hours after TBI (g). Femoral bone marrow was immunohistochemically stained for IL-12Rβ2 (orange color) 12 days after irradiation. While bone marrow from mice treated with vehicle lacked IL-12Rβ2–expressing cells and showed no signs of hematopoietic regeneration (b), mice treated with m-HemaMax showed hematopoietic reconstitution and the presence of IL-12Rβ2–expressing megakaryocytes, myeloid progenitors, and osteoblasts (c–f). Mice treated with HemaMax showed IL-12Rβ2–expressing osteoblasts but lacked megakaryocytes (g). Magnification = 100×.
Figure 6. Mice bone marrow hematopoietic stem…
Figure 6. Mice bone marrow hematopoietic stem cells, osteoblasts, and megakaryocytes express IL-12Rβ2.
Tissue sections obtained 30 days (a and c) and 12 days (b) after TBI (according to the protocol described in Figure 5) were stained immunohistochemically for IL-12Rβ2 (a and b, upper panels), markers of hematopoietic stem cells, Sca-1 (a, lower panel), and osteoblasts, osteocalcin (b, lower panel), or both IL-12Rβ2 and Sca-1 (c). Also both immature and mature megakaryocytes showed intense immunohistochemical staining for the presence of IL-12Rβ2 (c). Red arrows in (a) indicate hematopoietic stem cells that express IL-12Rβ2 while black arrows indicate those that do not express IL-12Rβ2. In IL-12Rβ2 and Sca-1 double staining (c) IL-12Rβ2 is stained pink while Sca-1 is stained brown. The subpopulation of stem cells co-expressing IL-12Rβ2 and Sca-1 as well as subpopulations expressing only IL-12Rβ2 or Sca-1 are indicated (c). Magnification = 100×.
Figure 7. m-HemaMax at low dose suppresses…
Figure 7. m-HemaMax at low dose suppresses radiation-induced intestinal injury in mice.
The IL-12Rβ2 expression in jejunal crypts (a) and the suppression of jejunal expression of LGR5 (b), a GI stem cell injury marker, are shown. Mice received vehicle (P5.6TT) or m-HemaMax subcutaneously at the indicated doses either in the absence of irradiation or 24 hours after TBI (8.6 Gy). Three days after irradiation, jejunum tissues were removed and immunohistochemically stained for IL-12Rβ2 (a) or LGR5 (b). Representative images show LGR5 in brown as indicated with arrows. Magnification = 400.
Figure 8. Similar exposures to m-HemaMax and…
Figure 8. Similar exposures to m-HemaMax and HemaMax at species-specific equivalent doses in mice and rhesus monkeys.
The plot of plasma AUClast of m-HemaMax versus the dose administered to mice in the absence of irradiation was linear at doses from 10 ng/mouse to 40 ng/mouse. The plasma AUClast of HemaMax at monkey equivalent doses of 20 ng/Kg and 80 ng/Kg was in good agreement with the extend of dose-dependent increases in m-HemaMax exposure in mice.
Figure 9. HemaMax administration increased plasma IFN-γ,…
Figure 9. HemaMax administration increased plasma IFN-γ, IL-18, EPO, IL-15, and neopterin concentrations in non-irradiated rhesus monkeys.
(a) Temporal kinetics of IFN-γ relative to that of HemaMax. (b) Temporal kinetics of IL-18 and EPO. (c) Temporal kinetics of IL-15 and neopterin. Animals received HemaMax subcutaneously at a dose of either 250 ng/Kg or 1000 ng/Kg in the absence of irradiation. The plasma concentrations of HemaMax, IFN-γ, IL-18, EPO, IL-15, and neopterin were determined by ELISA in blood samples withdrawn at the indicated times. n = 3 per timepoint in each group, except for neopterin, which was n = 1.
Figure 10. NHP and human bone marrow…
Figure 10. NHP and human bone marrow and small intestine express IL-12Rβ2.
Tissues from NHP and human femoral bone marrow (a) and jejunum/ileum (b) were immunohistochemically stained for IL-12Rβ2. (a) Progenitor cells and megakaryocytes expressing IL-12Rβ2 are shown. Adipocytes did not express IL-12Rβ2. (b) Intestinal crypts expressing IL-12Rβ2 are shown. Lymphoid cells in the lamina propria and submucosal regions also expressed IL-12Rβ2. C = crypt; LP = lamina propria. Magnification was 40× in (a) and 100× in (b).
Figure 11. HemaMax initiated at least 24…
Figure 11. HemaMax initiated at least 24 hours after irradiation increased percentage of survival of unsupported monkeys.
Individual dosing groups (a) and the pooled HemaMax dosing group (b) are shown. Animals were subjected to an LD50/30 of TBI at day 0 and subsequently received either vehicle (P5.6TT) or HemaMax subcutaneously at the indicated dosing regimens. Supportive care was prohibited during the study. Animals were monitored for survival up to 30 days. a One animal was excluded from the study due to a broken tooth.
Figure 12. HemaMax administration decreased leukopenia (a)…
Figure 12. HemaMax administration decreased leukopenia (a) and thrombocytopenia (b) at nadir in irradiated, unsupported rhesus monkeys.
Animals were subjected to an LD50/30 of TBI at day 0. Animals received subcutaneously either vehicle (P5.6TT) or HemaMax at a dose of 100 ng/Kg or 250 ng/Kg at 24 hours post TBI. Blood samples were withdrawn at the indicated times, and leukocytes and platelets were counted by an automated hematology analyzer.
Figure 13. Irradiated rhesus monkeys receiving HemaMax…
Figure 13. Irradiated rhesus monkeys receiving HemaMax had less body weights loss than animals receiving vehicle.
Body weights in Kg (a and b) and in percentage (c and d) are shown for the 100 ng/Kg and 250 ng/Kg dose groups. Monkeys were subjected to an LD50/30 of TBI at day 0 and subsequently received either vehicle (P5.6TT) or HemaMax subcutaneously at the indicated dosing regimens. Supportive care was prohibited during the study. Body weights were recorded every other day for up to day 30.
Figure 14. A multilevel model of HemaMax…
Figure 14. A multilevel model of HemaMax mechanism of action in increasing survival following exposure to radiation.
Current evidence suggests that HemaMax triggers responses at, at least, 4 levels in the body. At the Level 1 response, HemaMax promotes proliferation and activation of extant, radiosensitive immune cells, namely NK cells, macrophages, and dendritic cells. HemaMax-induced plasma elevations of IL-15 and IL-18 also facilitate maturation of NK cells, leading to the release of IFN-γ, which in turn, positively affects the production of endogenous IL-12 from macrophages and dendritic cells, and perhaps NK cells. These events enhance the innate immune competency early on following HemaMax administration. At the Level 2 response, HemaMax promotes proliferation and differentiation of the surviving hematopoietic stem cells, osteoblasts, and megakaryocytes into a specific cellular configuration that ensues optimal hematopoiesis. HemaMax-induced secretion of EPO from CD34+, IL-12Rβ2–positive bone marrow cells may also suppress local over-production of IFN-γ in the bone marrow and, thus, provide a milieu that promotes expansion of hematopoietic cells. Hematopoietic regeneration in the bone marrow enhances both innate and adaptive immune competency. At the Level 3 response, HemaMax preserves GI stem cells, leading to a reduction in pathogen leakage, an increase in food consumption, and a decrease in diarrhea. At the Level 4 response, HemaMax likely directly increases renal release of EPO, a cytoprotective factor, which enhances cellular viability in a diverse set of organs/tissues. Continued production of endogenous IL-12 primarily from dendritic cells activated by pathogens and/or EPO serves as a positive feedback loop and plays a key role in sustaining the initial response to exogenous HemaMax, perhaps for weeks after radiation. ↑ = increase; ↓ = decrease; HSC = Hematopoietic stem cells; NK cells = natural killer cells.

References

    1. Drouet M, Herodin F. Radiation victim management and the haematologist in the future: time to revisit therapeutic guidelines? Int J Radiat Biol. 2010;86:636–648.
    1. Donnelly EH, Nemhauser JB, Smith JM, Kazzi ZN, Farfan EB, et al. Acute radiation syndrome: assessment and management. South Med J. 2010;103:541–546.
    1. . Effects of radiation levels on human body. . Accessed June 19, 2011.
    1. Williams JP, Brown SL, Georges GE, Hauer-Jensen M, Hill RP, et al. Animal models for medical countermeasures to radiation exposure. Radiat Res. 2010;173:557–578.
    1. Johnson SM, Torrice CD, Bell JF, Monahan KB, Jiang Q, et al. Mitigation of hematologic radiation toxicity in mice through pharmacological quiescence induced by CDK4/6 inhibition. J Clin Invest. 2010;120:2528–36.
    1. Burdelya LG, Krivokrysenko VI, Tallant TC, Strom E, Gleiberman AS, et al. An agonist of toll-like receptor 5 has radioprotective activity in mouse and primate models. Science. 2008;320:226–30.
    1. Vijay-Kumar M, Aitken JD, Sanders CJ, Frias A, Sloane VM, et al. Flagellin treatment protects against chemicals, bacteria, viruses, and radiation. J Immunol. 2008;180:8280–5.
    1. Chen BJ, Deoliveira D, Spasojevic I, Sempowski GD, Jiang C, et al. Growth hormone mitigates against lethal irradiation and enhances hematologic and immune recovery in mice and nonhuman primates. PLoS One. 2010;5:e11056.
    1. Singh VK, Yadav VS. Role of cytokines and growth factors in radioprotection. Exp Mol Pathol. 2005;78:156–69.
    1. Hérodin F, Drouet M. Cytokine-based treatment of accidentally irradiated victims and new approaches. Exp Hematol. 2005;33:1071–80.
    1. Weiss JF, Landauer MR. History and development of radiation-protective agents. Int J Radiat Biol. 2009;85:539–73.
    1. Dumont F, Le Roux A, Bischoff P. Radiation countermeasure agents: an update. Expert Opin Ther Pat. 2010;20:73–101.
    1. Basile LA, Gallaher TK, Shibata D, Miller JD, Douer D. Multilineage hematopoietic recovery with concomitant antitumor effects using low dose Interleukin-12 in myelosuppressed tumor-bearing mice. J Transl Med. 2008;6:26.
    1. Chen T, Burke KA, Zhan Y, Wang X, Shibata D, et al. IL-12 facilitates both the recovery of endogenous hematopoiesis and the engraftment of stem cells after ionizing radiation. Exp Hematol. 2007;35:203–213.
    1. Colombo MP, Trinchieri G. Interleukin-12 in anti-tumor immunity and immunotherapy. Cytokine Growth Factor Rev. 2002;13:155–168.
    1. Jacobsen SE, Veiby OP, Smeland EB. Cytotoxic lymphocyte maturation factor (interleukin 12) is a synergistic growth factor for hematopoietic stem cells. J Exp Med. 1993;178:413–418.
    1. Bellone G, Trinchieri G. Dual stimulatory and inhibitory effect of NK cell stimulatory factor/IL-12 on human hematopoiesis. J Immunol. 1994;153:930–937.
    1. Ploemacher RE, van Soest PL, Boudewijn A, Neben S. Interleukin-12 enhances interleukin-3 dependent multilineage hematopoietic colony formation stimulated by interleukin-11 or steel factor. Leukemia. 1993;7:1374–1380.
    1. Ploemacher RE, van Soest PL, Voorwinden H, Boudewijn A. Interleukin-12 synergizes with interleukin-3 and steel factor to enhance recovery of murine hemopoietic stem cells in liquid culture. Leukemia. 1993;7:1381–1388.
    1. Hirayama F, Katayama N, Neben S, Donaldson D, Nickbarg EB, et al. Synergistic interaction between interleukin-12 and steel factor in support of proliferation of murine lymphohematopoietic progenitors in culture. Blood. 1994;83:92–98.
    1. Broxmeyer HE, Lu L, Platzer E, Feit C, Juliano L, et al. Comparative analysis of the influences of human gamma, alpha and beta interferons on human multipotential (CFU-GEMM), erythroid (BFU-E) and granulocyte-macrophage (CFU-GM) progenitor cells. J Immunol. 1983;131:1300–1305.
    1. Gimble JM, Medina K, Hudson J, Robinson M, Kincade PW. Modulation of lymphohematopoiesis in long-term cultures by gamma interferon: direct and indirect action on lymphoid and stromal cells. Exp Hematol. 1993;21:224–230.
    1. Means RT, Jr, Krantz SB. Inhibition of human erythroid colony-forming units by gamma interferon can be corrected by recombinant human erythropoietin. Blood. 1991;78:2564–2567.
    1. Terrell TG, Green JD. Comparative pathology of recombinant murine interferon-gamma in mice and recombinant human interferon-gamma in cynomolgus monkeys. Int Rev Exp Pathol. 1993;34 Pt B:73–101.
    1. Zoumbos NC, Djeu JY, Young NS. Interferon is the suppressor of hematopoiesis generated by stimulated lymphocytes in vitro. J Immunol. 1984;133:769–774.
    1. Kurz K, Gluhcheva Y, Zvetkova E, Konwalinka G, Fuchs D. Interferon-gamma-mediated pathways are induced in human CD34(+) haematopoietic stem cells. Immunobiology. 2010;215:452–457.
    1. Zhao X, Ren G, Liang L, Ai PZ, Zheng B, et al. Brief report: interferon-gamma induces expansion of Lin(−)Sca-1(+)C-Kit(+) Cells. Stem Cells. 2010;28:122–126.
    1. U.S. Food and Drug Administration. Center for Drug Evaluation and Research. FDA guidance estimating the maximum safe starting dose in initial clinical trials for therapeutics in adult healthy volunteers. 2005. . Accessed June 13, 2011.
    1. Yuan R, Maeda Y, Li W, Lu W, Cook S, et al. Erythropoietin: a potent inducer of peripheral immuno/inflammatory modulation in autoimmune EAE. PLoS One. 2008;3:e1924.
    1. Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 2007;449:1003–1007.
    1. Barker N, Huch M, Kujala P, van de Wetering M, Snippert HJ, et al. Lgr5(+ve) stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell. 2010;6:25–36.
    1. Garcia MI, Ghiani M, Lefort A, Libert F, Strollo S, et al. LGR5 deficiency deregulates Wnt signaling and leads to precocious Paneth cell differentiation in the fetal intestine. Dev Biol. 2009;331:58–67.
    1. Zou JJ, Schoenhaut DS, Carvajal DM, Warrier RR, Presky DH, et al. Structure-function analysis of the p35 subunit of mouse interleukin 12. J Biol Chem. 1995;270:5864–5871.
    1. Bekaii-Saab TS, Roda JM, Guenterberg KD, Ramaswamy B, Young DC, et al. A phase I trial of paclitaxel and trastuzumab in combination with interleukin-12 in patients with HER2/neu-expressing malignancies. Mol Cancer Ther. 2009;8:2983–2991.
    1. Lenzi R, Edwards R, June C, Seiden MV, Garcia ME, et al. Phase II study of intraperitoneal recombinant interleukin-12 (rhIL-12) in patients with peritoneal carcinomatosis (residual disease <1 cm) associated with ovarian cancer or primary peritoneal carcinoma. J Transl Med. 2007;5:66.
    1. Little RF, Aleman K, Kumar P, Wyvill KM, Pluda JM, et al. Phase 2 study of pegylated liposomal doxorubicin in combination with interleukin-12 for AIDS-related Kaposi sarcoma. Blood. 2007;110:4165–4171.
    1. Melichar B, Lenzi R, Rosenblum M, Kudelka AP, Kavanagh JJ, et al. Intraperitoneal fluid neopterin, nitrate, and tryptophan after regional administration of interleukin-12. J Immunother. 2003;26:270–276.
    1. Neta R, Stiefel SM, Finkelman F, Herrmann S, Ali N. IL-12 protects bone marrow from and sensitizes intestinal tract to ionizing radiation. J Immunol. 1994;153:4230–4237.
    1. Trinchieri G. Interleukin-12: a cytokine at the interface of inflammation and immunity. Adv Immunol. 1998;70:83–243.
    1. Langrish CL, McKenzie BS, Wilson NJ, de Waal MR, Kastelein RA, et al. IL-12 and IL-23: master regulators of innate and adaptive immunity. Immunol Rev. 2004;202:96–105.
    1. Gattoni A, Parlato A, Vangieri B, Bresciani M, Derna R. Interferon-gamma: biologic functions and HCV therapy (type I/II) (1 of 2 parts). Clin Ter. 2006;157:377–386.
    1. Macdougall IC, Cooper AC. Erythropoietin resistance: the role of inflammation and pro-inflammatory cytokines. Nephrol Dial Transplant. 2002;17(Suppl 11):39–43.
    1. Morceau F, Dicato M, Diederich M. Pro-inflammatory cytokine-mediated anemia: regarding molecular mechanisms of erythropoiesis. Mediators Inflamm. 2009;2009:405016.
    1. Dinarello CA, Fantuzzi G. Interleukin-18 and host defense against infection. J Infect Dis. 2003;187(Suppl 2):S370–S384.
    1. Gracie JA, Robertson SE, McInnes IB. Interleukin-18. J Leukoc Biol. 2003;73:213–224.
    1. Stonier SW, Schluns KS. Trans-presentation: a novel mechanism regulating IL-15 delivery and responses. Immunol Lett. 2010;127:85–92.
    1. Nakanishi K, Yoshimoto T, Tsutsui H, Okamura H. Interleukin-18 is a unique cytokine that stimulates both Th1 and Th2 responses depending on its cytokine milieu. Cytokine Growth Factor Rev. 2001;12:53–72.
    1. Werner ER, Werner-Felmayer G, Fuchs D, Hausen A, Reibnegger G, et al. Tetrahydrobiopterin biosynthetic activities in human macrophages, fibroblasts, THP-1, and T 24 cells. GTP-cyclohydrolase I is stimulated by interferon-gamma, and 6-pyruvoyl tetrahydropterin synthase and sepiapterin reductase are constitutively present. J Biol Chem. 1990;265:3189–3192.
    1. Fuchs D, Hausen A, Reibnegger G, Werner ER, Dierich MP, et al. Neopterin as a marker for activated cell-mediated immunity: application in HIV infection. Immunol Today. 1988;9:150–155.
    1. Anagnostou A, Liu Z, Steiner M, Chin K, Lee ES, et al. Erythropoietin receptor mRNA expression in human endothelial cells. Proc Natl Acad Sci U S A. 1994;91:3974–3978.
    1. Brines M, Cerami A. Emerging biological roles for erythropoietin in the nervous system. Nat Rev Neurosci. 2005;6:484–494.
    1. Buemi M, Cavallaro E, Floccari F, Sturiale A, Aloisi C, et al. The pleiotropic effects of erythropoietin in the central nervous system. J Neuropathol Exp Neurol. 2003;62:228–236.
    1. Fraser JK, Tan AS, Lin FK, Berridge MV. Expression of specific high-affinity binding sites for erythropoietin on rat and mouse megakaryocytes. Exp Hematol. 1989;17:10–16.
    1. Jaquet K, Krause K, Tawakol-Khodai M, Geidel S, Kuck KH. Erythropoietin and VEGF exhibit equal angiogenic potential. Microvasc Res. 2002;64:326–333.
    1. Sela S, Shurtz-Swirski R, Sharon R, Manaster J, Chezar J, et al. The polymorphonuclear leukocyte–a new target for erythropoietin. Nephron. 2001;88:205–210.
    1. Lifshitz L, Prutchi-Sagiv S, Avneon M, Gassmann M, Mittelman M, et al. Non-erythroid activities of erythropoietin: Functional effects on murine dendritic cells. Mol Immunol. 2009;46:713–721.
    1. Prutchi SS, Lifshitz L, Orkin R, Mittelman M, Neumann D. Erythropoietin effects on dendritic cells: potential mediators in its function as an immunomodulator? Exp Hematol. 2008;36:1682–1690.
    1. Cetin H, Olgar S, Oktem F, Ciris M, Uz E, et al. Novel evidence suggesting an anti-oxidant property for erythropoietin on vancomycin-induced nephrotoxicity in a rat model. Clin Exp Pharmacol Physiol. 2007;34:1181–1185.
    1. Genc S, Akhisaroglu M, Kuralay F, Genc K. Erythropoietin restores glutathione peroxidase activity in 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine-induced neurotoxicity in C57BL mice and stimulates murine astroglial glutathione peroxidase production in vitro. Neurosci Lett. 2002;321:73–76.
    1. Kumral A, Gonenc S, Acikgoz O, Sonmez A, Genc K, et al. Erythropoietin increases glutathione peroxidase enzyme activity and decreases lipid peroxidation levels in hypoxic-ischemic brain injury in neonatal rats. Biol Neonate. 2005;87:15–18.
    1. Liu J, Narasimhan P, Song YS, Nishi T, Yu F, et al. Epo protects SOD2-deficient mouse astrocytes from damage by oxidative stress. Glia. 2006;53:360–365.
    1. Wang ZY, Shen LJ, Tu L, Hu DN, Liu GY, et al. Erythropoietin protects retinal pigment epithelial cells from oxidative damage. Free Radic Biol Med. 2009;46:1032–1041.
    1. Akimoto T, Kusano E, Inaba T, Iimura O, Takahashi H, et al. Erythropoietin regulates vascular smooth muscle cell apoptosis by a phosphatidylinositol 3 kinase-dependent pathway. Kidney Int. 2000;58:269–282.
    1. Chong ZZ, Kang JQ, Maiese K. Erythropoietin is a novel vascular protectant through activation of Akt1 and mitochondrial modulation of cysteine proteases. Circulation. 2002;106:2973–2979.
    1. Chong ZZ, Kang JQ, Maiese K. Hematopoietic factor erythropoietin fosters neuroprotection through novel signal transduction cascades. J Cereb Blood Flow Metab. 2002;22:503–514.
    1. Parsa CJ, Matsumoto A, Kim J, Riel RU, Pascal LS, et al. A novel protective effect of erythropoietin in the infarcted heart. J Clin Invest. 2003;112:999–1007.
    1. Gerosa F, Baldani-Guerra B, Nisii C, Marchesini V, Carra G, et al. Reciprocal activating interaction between natural killer cells and dendritic cells. J Exp Med. 2002;195:327–333.
    1. Lodoen MB, Lanier LL. Natural killer cells as an initial defense against pathogens. Curr Opin Immunol. 2006;18:391–398.
    1. Varma TK, Lin CY, Toliver-Kinsky TE, Sherwood ER. Endotoxin-induced gamma interferon production: contributing cell types and key regulatory factors. Clin Diagn Lab Immunol. 2002;9:530–543.
    1. de Barros AP, Takiya CM, Garzoni LR, Leal-Ferreira ML, Dutra HS, et al. Osteoblasts and bone marrow mesenchymal stromal cells control hematopoietic stem cell migration and proliferation in 3D in vitro model. PLoS One. 2010;5:e9093.
    1. Ahmed N, Khokher MA, Hassan HT. Cytokine-induced expansion of human CD34+ stem/progenitor and CD34+CD41+ early megakaryocytic marrow cells cultured on normal osteoblasts. Stem Cells. 1999;17:92–99.
    1. Hamada T, Mohle R, Hesselgesser J, Hoxie J, Nachman RL, et al. Transendothelial migration of megakaryocytes in response to stromal cell-derived factor 1 (SDF-1) enhances platelet formation. J Exp Med. 1998;188:539–548.
    1. Hodohara K, Fujii N, Yamamoto N, Kaushansky K. Stromal cell-derived factor-1 (SDF-1) acts together with thrombopoietin to enhance the development of megakaryocytic progenitor cells (CFU-MK). Blood. 2000;95:769–775.
    1. Kiel MJ, Morrison SJ. Maintaining hematopoietic stem cells in the vascular niche. Immunity. 2006;25:862–864.
    1. Wang JF, Liu ZY, Groopman JE. The alpha-chemokine receptor CXCR4 is expressed on the megakaryocytic lineage from progenitor to platelets and modulates migration and adhesion. Blood. 1998;92:756–764.
    1. Dominici M, Rasini V, Bussolari R, Chen X, Hofmann TJ, et al. Restoration and reversible expansion of the osteoblastic hematopoietic stem cell niche after marrow radioablation. Blood. 2009;114:2333–2343.
    1. Savino R, Ciliberto G. A paradigm shift for erythropoietin: no longer a specialized growth factor, but rather an all-purpose tissue-protective agent. Cell Death Differ. 2004;11(Suppl 1):S2–S4.
    1. Zhang S, Wang Q. Factors determining the formation and release of bioactive IL-12: regulatory mechanisms for IL-12p70 synthesis and inhibition. Biochem Biophys Res Commun. 2008;372:509–512.
    1. Lacy MQ, Jacobus S, Blood EA, Kay NE, Rajkumar SV, et al. Phase II study of interleukin-12 for treatment of plateau phase multiple myeloma (E1A96): a trial of the Eastern Cooperative Oncology Group. Leuk Res. 2009;33:1485–1489.
    1. Lenzi R, Rosenblum M, Verschraegen C, Kudelka AP, Kavanagh JJ, et al. Phase I study of intraperitoneal recombinant human interleukin 12 in patients with Mullerian carcinoma, gastrointestinal primary malignancies, and mesothelioma. Clin Cancer Res. 2002;8:3686–3695.
    1. Leonard JP, Sherman ML, Fisher GL, Buchanan LJ, Larsen G, et al. Effects of single-dose interleukin-12 exposure on interleukin-12-associated toxicity and interferon-gamma production. Blood. 1997;90:2541–2548.
    1. Chao NJ. Accidental or intentional exposure to ionizing radiation: biodosimetry and treatment options. Exp Hematol. 2007;35:24–27.

Source: PubMed

3
Předplatit